Synthesis and Pharmacology of Conformationally Restricted

Timothy A. Grese,* Lewis D. Pennington, James P. Sluka, M. Dee Adrian, Harlan W. Cole, Tina R. Fuson,. David E. Magee, D. Lynn Phillips, Ellen R. Rowl...
0 downloads 0 Views 190KB Size
1272

J. Med. Chem. 1998, 41, 1272-1283

Synthesis and Pharmacology of Conformationally Restricted Raloxifene Analogues: Highly Potent Selective Estrogen Receptor Modulators Timothy A. Grese,* Lewis D. Pennington, James P. Sluka, M. Dee Adrian, Harlan W. Cole, Tina R. Fuson, David E. Magee, D. Lynn Phillips, Ellen R. Rowley, Pamela K. Shetler, Lorri L. Short, Murali Venugopalan, Na N. Yang, Masahiko Sato, Andrew L. Glasebrook, and Henry U. Bryant Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285 Received October 9, 1997

The 2-arylbenzothiophene raloxifene, 1, is a selective estrogen receptor modulator (SERM) which is currently under clinical evaluation for the prevention and treatment of postmenopausal osteoporosis. In vivo structure-activity relationships and molecular modeling studies have indicated that the orientation of the basic amine-containing side chain of 1, relative to the stilbene plane, is an important discriminating factor for the maintenance of tissue selectivity. We have constructed a series of analogues of 1 in which this side chain is held in an orientation which is orthogonal to the stilbene plane, similar to the low-energy conformation predicted for raloxifene. Herein, we report on the synthesis of these compounds and on their activity in a series of in vitro and in vivo biological assays reflective of the SERM profile. In particular, we describe their ability to (1) bind the estrogen receptor, (2) antagonize estrogen-stimulated proliferation of MCF-7 cells in vitro, (3) stimulate TGF-β3 gene expression in cell culture, (4) inhibit the uterine effects of ethynyl estradiol in immature rats, and (5) potently reduce serum cholesterol and protect against osteopenia in ovariectomized (OVX) rats without estrogen-like stimulation of uterine tissue. These data demonstrate that one of these compounds, LY357489, 4, is among the most potent SERMs described to date with in vivo efficacy on bone and cholesterol metabolism in OVX rats at doses as low as 0.01 mg/kg/d. Introduction The decreased production of ovarian steroids which occurs after the climacteric has been linked to a number of postmenopausal pathologies, particularly osteoporosis and coronary artery disease.1,2 Although estrogen replacement therapy (ERT) reduces the risks associated with these pathologies, concerns relating to the increased risk of endometrial cancer have necessitated the development of therapeutic regimens in which the uterine effects of estrogen are opposed by progestin treatment.3 Side effects of progestin treatment, such as resumption of menses, central nervous system disturbances, and the possibility of attenuated cardiovascular benefits, have significantly reduced patient compliance.4 Furthermore, recent studies which suggest an increased risk of breast and uterine cancer associated with estrogen replacement therapy have stimulated the search for treatment alternatives.5,6 Historically, a number of nonsteroidal compounds which interact with the estrogen receptor (ER) have been investigated as contraceptives and for the treatment of breast cancer, uterine dysfunction, and other disorders of the female reproductive system.7 Tamoxifen, originally developed as an estrogen antagonist and widely utilized for the treatment of breast cancer, has paradoxically been found to act as a partial estrogen agonist in the uterus and to display estrogen agonist effects in bone and in the cardiovascular system.8 More recently, selective estrogen receptor modulators (SERMs) which fully antagonize the effects of estrogen on uterine and mammary tissue, while mimicking the effects of estrogen on bone and the cardiovascular system, have

been investigated as a possible alternative to ERT.9 One such compound, raloxifene (LY139481 HCl; 1), is in advanced clinical trials for the prevention and treatment of osteoporosis.10,11

Pharmacologically, raloxifene is distinguished from tamoxifen by its lack of proliferative effects in uterine

S0022-2623(97)00688-2 CCC: $15.00 © 1998 American Chemical Society Published on Web 03/12/1998

Synthesis of Raloxifene Analogues

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8 1273

Figure 1. Overlay of minimized structures of raloxifene (green carbons) and 3a (gray carbons).

Scheme 1a

a Reagents: (a) HCl, H O, THF, reflux; (b) H O , Na CO (see ref 24); (c) Et N, EtOH, CH Cl ; (d) DDQ, dichloroethane, 80 °C; (e) 2 2 2 2 3 3 2 2 AlCl3, EtSH, CH2Cl2; (f) TBDMSCl, Et3N, CH2Cl2; (g) NaH, (MeO)2CO, MeOH (see ref 25); (h) resorcinol, P(O)Cl3, toluene, 80 °C.

tissue.12,13 Other researchers have developed a series of benzopyran-containing molecules (e.g. EM-800,14 CDRI-85/28715) which also exhibit this tissue specificity.16 We have recently speculated that this difference in biological activity may be related to a specific structural property, that is, the orientation of the basic amine-containing side chain with respect to the stilbene plane.16 While in tamoxifen this side chain is coplanar with the stilbene moiety, both molecular modeling and X-ray crystallographic studies predict an orthogonal orientation for the side chain in raloxifene (Figure 1).17 We have demonstrated that raloxifene analogues which are forced to adopt the tamoxifen orientation exhibit a tamoxifen-like biological profile, while analogues which are conformationally locked in a raloxifene-like orientation maintain the raloxifene profile.18 Herein, we describe the synthesis and biological activity of a series of conformationally locked ER modulators which incorporate structural elements of both raloxifene and the aforementioned benzopyrans. In particular, we disclose LY357489, 4, an extremely potent SERM with in vivo efficacy on bone and cholesterol metabolism at doses as low as 0.01 mg/kg. Chemistry Compounds 2-4 were designed on the basis of molecular modeling studies of raloxifene and benzopyrancontaining SERMs which predicted an orthogonal orientation of their respective side chains relative to the stilbene plane.16 Overlay of the minimized structure of compound 3 with raloxifene (Figure 1) indeed predicted a similar topology.19,20 A retrosynthetic disconnection of the side chain of compounds 2-4, leading to the tetracyclic coumarins

5-7 and a functionalized aryl Grignard reagent, 22, allowed the utilization of methodology previously developed for the synthesis of 2,3-diarylisoflav-3-enes.21 Compound 5 was readily available by the silylation of commercially available coumestrol.22 Thiacoumestans 6a-c were prepared by the condensation of a salicyl aldehyde with thianaphthen-2-ones 10a,b followed by oxidation, demethylation, and silylation (Scheme 1a).23 This protecting group interchange was necessary, both for ease of removal and to provide improved solubility in later stages of the synthesis (vide supra). Compounds 10a,b were in turn prepared either by hydrolysis of the 2-(dimethylamino)benzothiophene, 8,24,25 or from benzothiophene via oxidation of the corresponding boronic acid, 9.26 Naphthocoumarin 7 was prepared from carbomethoxytetralone 1227 via a von Pechman condensation,28 followed by protecting group interchange and dehydrogenation (Scheme 1b). Reduction of the tetracyclic coumarins 5-7 with diisobutylaluminum hydride (DIBAL-H) then provided the corresponding lactols 14-16 (Scheme 2).21 The silyl protecting groups were required at this stage, since the reduction was best carried out in toluene or toluene/ dichloromethane mixtures at low temperatures and the corresponding methoxy-substituted coumarins had limited solubility under these conditions. In the benzofuran-containing system, the lactol was isolated as a mixture with the corresponding aldehyde tautomer 17, and the yield of the reduction was significantly lower. Presumably, the ring strain engendered by the incorporation of the smaller furan ring leads to a larger proportion of the open form, which is prone to overreduction under the reaction conditions. For the benzothiophene-containing system, significant amounts of

1274

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8

Grese et al.

Scheme 2a

a Reagents: (a) DIBAL-H, toluene, -100 to -70 °C; (b) phenol, (MgSO ), chlorobenzene or CH Cl , room temperature to reflux; (c) 4 2 2 THF, 0 °C to room temperature; (d) TBAF, THF; (e) PCC, Celite, CH2Cl2.

starting material and overreduced product, 18, were also obtained. Diol 18 could be recycled to 6 by oxidation with PCC; however, partial oxidation to the lactol 16 was not feasible. Conversion of the lactols to the corresponding phenyl acetals 19-21 and displacement with the aryl Grignard reagent 22 proceeded as expected, and desilylation then provided the desired compounds 2-4 in racemic form.21 Alternatively, condensation of 20a with (4-((trimethylsilyl)oxy)phenyl)magnesium bromide29 followed by selective removal of the trimethylsilyl moiety provided monophenol 23 (Scheme 3). Mitsunobu alkylation with a variety of N-hydroxyethylamines followed by desilylation then provided analogues 3f-j containing modified side chains. Finally, deshydroxy analogue 3d was prepared from 3a by conversion to the bistriflate and palladium-catalyzed transfer hydrogenation, and dimethoxy analogue 3e was prepared by methylation of 3a with diazomethane (Scheme 4).

Scheme 3a

Biological Testing

a Reagents: (a) (i) [4-[(trimethylsilyl)oxy]phenyl]magnesium bromide, THF; (ii) K2CO3, MeOH; (b) HOCH2CH2Y, PPh3, DEAD, toluene; (c) TBAF, THF.

In Vitro. ER binding affinities were determined by displacement of bound [3H]-17β-estradiol from MCF-7 cell lysate for compounds 2-4 and are reported in Table 1.30 Antagonism of estrogen action in a mammary tumor cell line was assayed via inhibition of MCF-7 cell proliferation stimulated by 10-11 M 17β-estradiol and IC50 values are also included in Table 1.31 In Vivo. Tissue-specific estrogen agonist effects were examined in OVX rats,13 utilizing uterine weight, uterine eosinophil peroxidase (EPO) activity,32 and serum cholesterol levels as endpoints after 4 days of

treatment (Table 1, Figure 2). Selected compounds were further evaluated in a 5-week, OVX rat model in which effects on bone mineral density (BMD) were also examined.33 Bone-mineral density was assessed at the proximal aspect of the tibia by quantitative computed tomography (QCT) and is reported as percent protection relative to sham-operated and OVX controls (Figure 3). In vivo estrogen antagonist activity in the uterus was determined in 21-day-old female Sprague-Dawley rats dosed with a maximally stimulatory dose of 17R-

Synthesis of Raloxifene Analogues

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8 1275

Scheme 4a

raloxifene also stimulates a modest increase in uterine wet weight, this increase is not dose related and is not coincident with increases in other measures of uterine hypertrophy such as epithelial cell height and total estrogenicity.12a This uterine weight increase has therefore been attributed to water retention.12b The inconsistent correlation between elevation of uterine weight and stimulation of other uterine parameters has led to reliance on uterine EPO activity, a very sensitive indicator of estrogen action in the uterus.32 Although ethynylestradiol and tamoxifen both potently reduce serum cholesterol, they also induce significant increases in uterine EPO while raloxifene does not. All three compounds induce significant increases in uterine weight, although the effect of raloxifene is relatively modest and is not dose related. We have used uterine EPO, therefore, to discriminate compounds which demonstrate a tamoxifen-like profile from those whose activity parallels that of raloxifene.34,35 As shown in Table 1, the conformationally restricted SERMs generally mimic the effects of raloxifene on uterine parameters and serum cholesterol. In contrast to original the raloxifene series, in which the deshydroxy congeners showed increased in vivo potency, changes in the hydroxylation pattern of the benzothienobenzopyran (3a-d) resulted in a reduction in potency.33 This distinction may reflect different efficiency or selectivity of metabolic hydroxylation for the two series. Similar to the raloxifene series,18 changing the base from piperidine to other cyclic or acyclic amine functionality reduced both potency and selectivity, with 3f-j all showing increased evidence of uterine stimulation relative to 3a.38 The piperidine-containing compounds 2, 3a-c, and 4 produce no significant increase in uterine EPO or, unlike raloxifene, uterine weight. A similar lack of uterine weight effects has previously been reported for another series of benzopyran-containing SERMs, but the relevance of this difference in comparison with raloxifene is unknown.16 Figure 2 demonstrates the surprising increase in in vivo potency which was observed across the benzofuran (2), benzothiophene (3a), naphthalene (4) series. Although similar to 3a and raloxifene in vitro, compound 4 (LY357489) demonstrated remarkable potency (ED50 ) 0.02 mg/kg/d) for lowering serum cholesterol in the OVX rat. Although the reasons for this increased potency relative to 3a are unclear, other highly potent, naphthalene-containing SERMs have been described previously.37,39 Compounds 3a and 4 were further evaluated in an OVX rat model of estrogen-deficiency-induced osteopenia. As shown in Figure 3, both 3a and 4 maintain the ability of raloxifene and estrogen to prevent ovariectomy-induced bone loss. In particular, LY357489, 4, showed increased potency relative to raloxifene, with significant effects at doses as low as 0.01 mg/kg/d. Compounds 2, 3a, and 4 were also evaluated for uterine estrogen antagonist activity in the immature rat model as shown in Figure 4. In this model also, LY357489, 4, showed improved potency (ED50 ) 0.05 mg/kg/d, raloxifene ED50 ) 0.55 mg/kg/d) and was even able to reduce uterine weight below the level observed for untreated controls. Compound 3a again showed

a Reagents: (a) PhN(Tf) , Et N, DMF; (b) Pd(PPh ) , dppp, 2 3 3 4 HCO2H, Et3N, DMF; (c) CH2N2, Et2O, MeOH.

ethynylestradiol (0.1 mg/kg) for 3 consecutive days.34 Test groups were also administered various doses of compounds 2-4 15 min prior to ethynylestradiol dosing for 3 days. Uterine weight/body weight ratios were calculated for each animal and are reported as percent inhibition (Figure 4). Results In Vitro. The in vitro results in Table 1 indicate the importance of the hydroxy functionality of 2-4 for optimal ER binding and inhibition of MCF-7 cell proliferation. Comparison of the binding activities of 3b-e with that of 3a reveals 6-20-fold decreases in relative binding affinity (RBA) when either or both of the hydroxy groups are replaced with a proton or converted to methyl ethers. In parallel fashion, MCF-7 inhibitory potency is decreased by 30-500-fold for the same compounds. A similar importance has previously been reported in the raloxifene series; however, unlike the raloxifene series in which the 6-hydroxy functionality is clearly more important for in vitro biological activity, compounds 3b and 3c show only minor differentiation.35 The importance of the nature of the amine substituents with respect to in vitro estrogen antagonism is demonstrated by the compounds 3f-j and is similar to effects which have been reported in the raloxifene series, with the piperidine base providing the optimum activity.18 Interestingly, the amine substituent has little effect on the receptor binding observed for these compounds and yet decreases MCF-7 inhibitory potency 3-50-fold relative to that observed for 3a. Finally, the replacement of the benzothiophene substructure of 3a with a benzofuran (2) or a naphthalene (4) produces relatively minor effects on the in vitro biological activity. The lower binding affinity observed with 2 may be ascribed to the general intolerance of the ER toward polar substitution.36 The replacement of the benzothiophene core of raloxifene with a naphthalene has previously been shown to provide compounds with a similar pharmacological profile.37 In Vivo. The in vivo data from the 4-day OVX rat assay (Table 1, Figure 2) provide additional insights into the effects of structural modifications on the complex biological activities of SERMs. For example, tamoxifen stimulates a dose dependent increase in uterine wet weight as well as increases in other measures of uterine stimulation such as epithelial cell height.12a Although

1276

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8

Grese et al.

Table 1. In Vitro and in Vivo Biological Activity of Conformationally Restricted SERMs

a RBA ) relative binding affinity by competition with [3H]-17β-estradiol. b Average of at least two determinations. Values are (10%. Dose required to give 50% inhibition of a maximally effective (10-11) dose of 17β-estradiol. Average of at least three determinations. Values are (10%. d MED ) minimally effective dose (mg/kg body weight) at which a statistically significant (p e 0.05) increase in uterine weight/body weight was observed. Activity at the MED is expressed as percent increase relative to OVX controls ( standard error. e MED at which a significant (>5-fold increase relative to OVX control and value of Vmax g 10) increase in EPO activity was observed. Activity at the MED is expressed as Vmax ( standard error. f Percent decrease in serum cholesterol relative to OVX controls ( standard error. Statistically significant (p e 0.05) differences are denoted by an asterisk (*). g Dose required to reduce serum cholesterol by 50% relative to OVX controls. h NA ) not active at the doses tested. i 4-Hydroxytamoxifen, the primary biologically active metabolite of tamoxifen. j Not determined. k Maximum dose tested ) 1.0 mg/kg. l Maximum dose tested ) 3.0 mg/kg. c

potency similar to that of raloxifene, while compound 2 was somewhat less effective. Discussion As part of our program to explore structure-activity relationships in the raloxifene series, we discovered a unique dependence upon the orientation of the basic amine-containing side chain for the maintenance of full estrogen antagonist activity in the uterus.16,18 Herein, we have exploited this finding in the design of a new series of SERMs in which the side chain is rigidly held in an orthogonal orientation relative to a tetracyclic, steroid-like core. Indeed, as described above, these compounds function as full antagonists in the OVX rat uterus without any evidence of intrinsic agonist activity in that tissue. Nevertheless, as with raloxifene, they maintain the ability to reduce serum cholesterol and protect against ovariectomy-induced osteopenia. The

structure-activity relationship observed in the benzothiophene-containing series, 3, nicely parallels that reported for raloxifene,18,35 although there is increased dependence upon the degree of hydroxylation for in vivo, as well as in vitro, potency. Surprisingly, substitution of oxygen or ethylene for the benzothiophene sulfur revealed a marked dependence of in vivo potency upon the nature of X (Scheme 1), with 4 (X ) CH2dCH2) > 3a (X ) S) > 2 (X ) O), roughly correlated with increasing hydrophobicity of the steroid-like nucleus. Most notably, shifting from the benzothiophene to the naphthalene-containing nucleus of 4 resulted in a significant boost in in vivo potency of both agonist and antagonist activities, without a corresponding increase in in vitro effects. This shift may reflect improved bioavailability for 4, although the reasons for this improvement are unclear. Compound 4 maintains the hydroxylation pattern of 3a and ral-

Synthesis of Raloxifene Analogues

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8 1277

Figure 2. Effects of 2, 3a, and 4 on serum cholesterol levels in OVX rats. Cholesterol values are reported as the mean percent decrease relative to OVX control ((SEM) with n g 5 for all groups. Raloxifene- and ethynylestradiol-treated groups are included as internal standards. Observed effects at doses of 2 g 1.0 mg/kg/d, 3a g 0.1 mg/kg/d, 4 g 0.01 mg/kg/d and all doses of raloxifene or ethynyl estradiol differ significantly from OVX control at P e 0.05.

Figure 3. Effects of raloxifene, 3a, and 4 on bone mineral density (BMD) in the OVX rat. BMD values are reported as percent protection ((SEM) relative to OVX controls, with sham control values defined as 100% and OVX controls defined as 0 and n g 6 for each group. Observed effects at doses of 3a or raloxifene g 1.0 mg/kg/d and doses of 4 or ethynyl estradiol g 0.01 mg/kg/d differ significantly from OVX control at P e 0.05.

oxifene, and glucuronidation of the phenolic moieties is thought to be the primary factor limiting the bioavailability of raloxifene.40 Nevertheless, improved bioavailability has been recently claimed for another series of hydroxylated SERMs which are built upon a tetrahydronaphthalene core unit.39 Further experimentation will be necessary to determine if pharmacokinetics are indeed responsible for this effect. One alternative explanation involves the mechanisms by which the ER controls gene transcription. It has recently been proposed that the conformation of the ER-ligand complex is dependent upon the nature of the interacting ligand and that the distinct conformation induced by a particular ligand affects differentially which genes are transcriptionally activated.41,42 To the extent that structural modification of the ligand disrupts or alters this receptor-ligand conformation, different sets of estrogen responsive genes may be influenced positively or negatively, resulting in an altered biological profile. We have explored the effects of LY357489, 4, on TGF-β3 promoter activity in vitro, a candidate gene which may be involved in the bone effects of raloxifene, and found no significant increase in potency (Figure 5).43 Nevertheless, further evaluation of the effects 4 at the transcriptional level in other promoter systems may be warranted. The compounds examined within this study were prepared in racemic form, and therefore the biological results reported herein must be interpreted with caution. The possibility that the individual enantiomers may exert differential biological activities, functioning synergistically in some tissues and antagonistically in

others, cannot be excluded. Studies with other chiral, nonsteroidal estrogen receptor modulators have indicated that although the majority of the biological effects reside in a single enantiomer, the racemates exhibit the same qualitative profile of activity.14,44 Further studies elucidating the biological effects of individual enantiomers of 4 will be reported in due course. In conclusion, we have prepared a series of novel raloxifene analogues in which the side chain is constrained in an orientation which approximates the lowenergy conformation predicted for the parent molecule. We have demonstrated that this series of compounds maintain the SERM profile of biological activity which has previously been described for raloxifene, including ER binding, inhibition of MCF-7 cell proliferation, estrogen antagonist effects in the immature rat uterus, and tissue-specific estrogen agonist effects on serum cholesterol and bone in the OVX rat.12b,34 Finally, we have demonstrated that one of these compounds, LY357489, 4, is among the most potent SERMs described to date with in vivo efficacy on bone and cholesterol metabolism in OVX rats at doses as low as 0.01 mg/kg/d. Experimental Section Chemistry. General experimental procedures have been recently published.35 The abbreviations THF, DMF, DCE, DMSO, DDQ, TBAF, and DEAD refer to tetrahydrofuran, dimethylformamide, 1,2-dichloroethane, dimethyl sulfoxide, 2,3-dichloro-5,6-dicyano-1,4-benzoquinone, tetra-n-butylammonium fluoride, and diethyl azodicarboxylate, respectively. All spectra were recorded in acetone-d6 unless otherwise indicated. Elemental analyses were carried out by the Physi-

1278

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8

Figure 4. Effect of raloxifene, 2, 3a, and 4 on ethynylestradiol (EE)-induced uterine weight increase in immature rats. Data are reported as percent inhibition of uterine weight/body weight ((SEM) relative to EE-treated controls, with vehicle control values defined as 100% and EE-treated controls as 0 and n g 6 for each group. Observed effects at doses of 2 g 1.0 mg/kg/d and doses of 3a, 4, and raloxifene g 0.1 mg/kg/d differ significantly from EE-treated controls at P e 0.05.

Figure 5. Effects of raloxifene, 1, and LY357489, 4, on TGFβ3 promoter activity. Data are reported as the mean foldinduction ((SEM) versus matched controls with n ) 3 for all groups. cal Chemistry Department of Lilly Research Laboratories and are within (0.4% of theory unless otherwise noted. 6-Methoxythianaphthen-2-one (10a).25 A solution of 824 (75.1 g, 360 mmol) in THF (600 mL) was treated with 1 N

Grese et al. HCl (615 mL) and heated at reflux for 12 h. The mixture was neutralized with saturated NaHCO3, diluted with ether (1 L), washed with saturated NaHCO3 (2 × 1 L) and brine (1 L), dried (MgSO4), and concentrated. The residue was recrystallized from ether to provide 52.7 g (81%) of the title compound: 1 H NMR (CDCl3) δ 7.16 (d, J ) 9.7 Hz, 1H), 6.87 (d, J ) 2.9 Hz, 1H), 6.73 (dd, J ) 9.7, 2.9 Hz, 1H), 3.88 (s, 2H), 3.79 (s, 3H). (()-6a,11a-Dihydro-3,9-dimethoxy-6H-[1]benzothieno[3,2-c][1]benzopyran-6-one (11a). To a stirred solution of 10a (20 g, 111 mmol) in a mixture of EtOH (100 mL) and CH2Cl2 (50 mL) was added 4-methoxysalicyl aldehyde (17.5 g, 115 mmol) followed by Et3N (567 mg, 784 mL, 5.6 mmol) at room temperature. After 30 min, a solid began to precipitate, and stirring was continued overnight. The mixture was then diluted with cold hexane (1 L) and filtered to yield 28.7 g (82%) of the title product as an off-white solid, pure by 1H NMR analysis. An analytical sample was obtained by recrystallization from toluene as light yellow crystals, mp 157-165 °C (dec): 1H NMR (CDCl3) δ 7.33 (d, J ) 8.6 Hz, 1H), 7.31 (d, J ) 8.1 Hz, 1H), 6.75 (d, J ) 2.4 Hz, 1H), 6.70 (m, 2H), 6.60 (d, J ) 2.5 Hz, 1H), 5.22 (d, J ) 7.2 Hz, 1H), 4.33 (d, J ) 7.2 Hz, 1H), 3.79 (s, 3H), 3.76 (s, 3H); IR (CHCl3) 1759 cm-1; MS (FD) m/e 314 (M+). Anal. (C17H14O4S) C, H. (()-6a,11a-Dihydro-9-methoxy-6H-[1]benzothieno[3,2c][1]benzopyran-6-one (11c). The title compound was prepared in 83% yield from 10a and salicyl aldehyde by a method similar to that described for 11a: 1H NMR (CDCl3) δ 7.45 (d, J ) 7.5 Hz, 1H), 7.33 (m, 2H), 7.17 (m, 1H), 7.07 (d, J ) 8.1 Hz, 1H), 6.77 (d, J ) 2.4 Hz, 1H), 6.69 (dd, J ) 2.4, 8.5 Hz, 1H), 5.25 (d, J ) 7.4 Hz, 1H), 4.37 (d, J ) 7.3 Hz, 1H), 3.77 (s, 3H); 13C NMR (CDCl3) δ 166.4, 160.7, 150.8, 141.4, 130.1, 129.0, 127.7, 127.0, 124.9, 119.5, 117.2, 111.2, 108.3, 55.5, 50.6, 49.6; IR (CHCl3) 1766 cm-1; MS (FD) m/e 284 (M+). Anal. (C16H12O3S) C, H. 3,9-Bis[(tert-butyldimethylsilyl)oxy]-6H-[1]benzothieno[3,2-c][1]benzopyran-6-one (6a). A mixture of 11a (4.5 g, 14.3 mmol) and DDQ (3.4 g, 15 mmol) in DCE (100 mL) was heated briefly to 80 °C, inducing the formation of a precipitate. The mixture was filtered hot; the precipitate was rinsed with CH2Cl2 and the mother liquor concentrated in vacuo. The remnant was dissolved in hot CH2Cl2, filtered to remove residual hydroquinone, and reconcentrated. The product was recrystallized from toluene to provide 3.94 g (88%) of 9-methoxy-6H-[1]benzothieno[3,2-c][1]benzopyran-6-one as white needles, mp 220-221 °C: 1H NMR (acetone-d6/DMSO-d6) δ 8.41 (d, J ) 8.9 Hz, 1H), 7.82 (d, J ) 8.7 Hz, 1H), 7.74 (d, J ) 2.3 Hz, 1H), 7.20 (dd, J ) 8.9, 2.4 Hz, 1H), 7.09 (d, J ) 2.4 Hz, 1H), 7.03 (dd, J ) 8.6, 2.4 Hz, 1H), 3.94 (s, 3H), 3.91 (s, 3H); IR (KBr) 1717 cm-1; MS (FD+) m/e 312 (M+). To a mechanically stirred slurry of the product prepared above (12 g, 38.4 mmol) in CH2Cl2 (220 mL) was added ethanethiol (11.9 g, 13.4 mL, 192 mmol) followed by aluminum chloride (38.4 g, 288 mmol), portionwise. The reaction mixture was stirred at ambient temperature for 5 h, cooled to 0 °C, and quenched cautiously with THF (250 mL) followed by saturated NaHC3 (250 mL). The mixture was diluted with THF (1 L), the layers were separated, and the aqueous layer was washed with THF (200 mL). The combined organic layers were dried (Na2SO4) and concentrated to yield 11.1 g of crude diphenol as a yellow solid, which was used without further purification. The crude product was slurried in CH2Cl2 (220 mL) and treated with Et3N (20.2 g, 28 mL, 200 mmol) and tertbutyldimethylsilyl chloride (20.3 g, 134.4 mmol). The mixture was stirred at ambient temperature for 5 h, during which it slowly became homogeneous. After dilution with hexane (600 mL), the mixture was washed with brine (600 mL) and the aqueous layer was extracted with hexane (300 mL). The combined organic layers were dried (Na2SO4) and concentrated, and the residue was recrystallized from hexane to provide 18.0 g (91%) of the title compound as a fluffy white solid, mp 142-144 °C: 1H NMR (CDCl3) δ 8.50 (d, J ) 8.7 Hz, 1H), 7.58 (d, J ) 8.5 Hz, 1H), 7.32 (d, J ) 2.1 Hz, 1H),

Synthesis of Raloxifene Analogues

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8 1279

7.05 (dd, J ) 8.7, 2.1 Hz, 1H), 6.92 (d, J ) 2.2 Hz, 1H), 6.84 (dd, J ) 8.4, 2.2 Hz, 1H), 1.01 (s, 9H), 1.00 (s, 9H), 0.27 (s, 6H), 0.25 (s, 6H); 13C NMR (DMSO-d6) δ 158.4, 157.0, 154.6, 152.7, 149.6, 138.5, 130.5, 125.2, 124.6, 120.0, 117.6, 116.3, 112.7, 111.3, 108.0, 25.6, 25.6, 18.2, 18.2, -4.4, -4.4; IR (CHCl3) 1717 cm-1; MS (FD) m/e 512 (M+). Anal. (C27H36O4SSi2) C, H. 3-[(tert-Butyldimethylsilyl)oxy]-6H-[1]benzothieno[3,2-c][1]benzopyran-6-one (6b). The title compound was prepared from 6a,11a-dihydro-3-methoxy-6H-[1]benzothieno[3,2-c][1]benzopyran-6-one (11b)23 in three steps and 61% overall yield by a method similar to that described for 6a: 1H NMR δ 8.57 (d, J ) 7.8 Hz, 1H), 8.10 (d, J ) 8.0 Hz, 1H), 7.81 (d, J ) 8.2 Hz, 1H), 7.5-7.7 (m, 2H), 6.99 (m, 2H), 1.03 (s, 9H), 0.33 (s, 6H); IR (CHCl3) 1716 cm-1; MS (FD) m/e 382 (M+). Anal. (C21H22O3SSi) C, H. 9-[(tert-Butyldimethylsilyl)oxy]-6H-[1]benzothieno[3,2-c][1]benzopyran-6-one (6c). The title compound was prepared from 11c in three steps and 73% overall yield by a method similar to that described for 6a: 1H NMR (CDCl3) δ 8.55 (d, J ) 8.5 Hz, 1H), 7.72 (d, J ) 8.7 Hz, 1H), 7.4-7.6 (m, 2H), 7.34 (m, 2H), 7.08 (dd, J ) 2.2, 8.8 Hz, 1H), 1.02 (s, 9H), 0.26 (s, 6H); IR (CHCl3) 1717 cm-1; MS (FD) m/e 382 (M+). Anal. (C21H22O3SSi) C, H. (()-3,9-Bis[(tert-butyldimethylsilyl)oxy]-6H-[1]benzothieno[3,2-c][1]benzopyran-6-ol (15a). A solution of 6a (2.0 g, 3.9 mmol) in toluene (200 mL) was cooled to -92 °C and treated dropwise with a 1.0 M toluene solution of diisobutylaluminum hydride (4.7 mL, 4.7 mmol) at a rate that maintained the internal temperature below -89 °C. The mixture was stirred for approximately 3 h as the temperature gradually rose to -77 °C and then was quenched with MeOH (5 mL) and 10% aqueous citric acid (50 mL). After dilution with CH2Cl2 (200 mL), the mixture was washed with saturated potassium sodium tartrate (100 mL), and the aqueous layer was extracted with CH2Cl2 (2 × 200 mL). The combined organic layers were washed with brine (300 mL), and the brine wash was further extracted with CH2Cl2 (100 mL). The organic layers were dried (Na2SO4) and concentrated, and the remnant was chromatographed (silica gel, 1-15% EtOAc/ hexane) to yield 360 mg (18%) of starting material, 1.21 g (60%, 74% based on recovered starting material), of the titled compound as a white crystalline solid (analytical sample recrystallized from hexane/EtOAc, mp 162-164 °C) [1H NMR δ 7.70 (d, J ) 9.0 Hz, 1H), 7.46 (d, J ) 2.1 Hz, 1H), 7.29 (d, J ) 8.1 Hz, 1H), 7.00 (dd, J ) 8.5, 1.8 Hz, 1H), 6.85 (br s, 1H), 6.6 (m, 2H), 6.39 (br s, 1H), 1.01 (s, 9H), 1.00 (s, 9H), 0.25 (s, 6H), 0.25 (s, 6H); 13C NMR δ 156.9, 153.3, 151.7, 139.9, 131.9, 124.5, 123.9, 121.7, 118.8, 113.7, 113.1, 112.7, 108.7, 90.7, 25.1, 25.1, 17.9, -5.2, -5.2; IR (CHCl3) 3540 cm-1; MS (FD+) m/e 514 (M+). Anal. (C27H38O4SSi2) C, H] and 260 mg (13%, 16% based on recovered starting material) of diol 18 as an amorphous solid: 1H NMR (CDCl3) δ 7.73 (d, J ) 8.7 Hz, 1H), 7.29 (d, J ) 2.0 Hz, 1H), 7.16 (d, J ) 8.2 Hz, 1H), 6.98 (dd, J ) 8.7, 2.0 Hz, 1H), 6.5 (m, 3H), 6.30 (br s, 1H), 4.71 (s, 2H), 1.01 (s, 9H), 1.00 (s, 9H), 0.22 (s, 6H), 0.19 (s, 6H); IR (CHCl3) 3600, 3510 cm-1; MS (FD+) m/e 516 (M+). (()-3-[(tert-Butyldimethylsilyl)oxy]-6H-[1]benzothieno[3,2-c][1]benzopyran-6-ol (15b). The title compound was prepared in 45% yield from 6b by a method similar to that described for 15a: 1H NMR δ 7.96 (d, J ) 7.7 Hz, 1H), 7.83 (d, J ) 6.7 Hz, 1H), 7.3-7.8 (m, 3H), 6.91 (m, 1H), 6.62 (m, 2H), 1.00 (s, 9H), 0.26 (s, 6H); IR (CHCl3) 2958, 2932, 2861, 1616, 1594 cm-1; MS (FD) m/e 384 (M+). Anal. (C21H24O3SSi) C, H. (()-9-[(tert-Butyldimethylsilyl)oxy]-6H-[1]benzothieno[3,2-c][1]benzopyran-6-ol (15c). The title compound was prepared in 49% yield from 6c by a method similar to that described for 15a: 1H NMR δ 7.77 (d, J ) 8.6 Hz, 1H), 7.50 (d, J ) 2.1 Hz, 1H), 7.42 (m, 1H), 7.28 (m, 1H), 7.05 (m, 2H), 6.89 (m, 1H), 6.46 (m, 1H), 1.02 (s, 9H), 0.26 (s, 6H); IR (CHCl3) 2959, 2932, 2861, 1612, 1598 cm-1; MS (FD) m/e 384 (M+). Anal. (C21H24O3SSi) C, H.

(()-3,9-Bis[(tert-butyldimethylsilyl)oxy]-6-phenoxy6H-[1]benzothieno[3,2-c][1]benzopyran (20a). To a solution of 15a (4.52 g, 8.78 mmol) and phenol (4.13 g, 43.9 mmol) in CH2Cl2 (100 mL) was added anhydrous MgSO4 (4.5 g), and the resultant slurry was stirred for 4 h at ambient temperature. The mixture was filtered and concentrated and the residue dissolved in chlorobenzene and reconcentrated in vacuo at approximately 70 °C. The residue was then dissolved in CH2Cl2 (300 mL), washed with saturated Na2CO3 (3 × 300 mL) and water (2 × 300 mL), dried (Na2SO4), and concentrated to yield 5.16 g (99%) of the title compound as an amorphous solid which was used without further purification: 1H NMR δ 7.67 (d, J ) 8.7 Hz, 1H), 7.50 (d, J ) 2.1 Hz, 1H), 7.4-7.5 (m, 4H), 7.23 (d, J ) 8.4 Hz, 1H), 7.08 (t, J ) 7.2 Hz, 1H), 6.99 (dd, J ) 8.7, 2.1 Hz, 1H), 6.67 (dd, J ) 8.4, 2.3 Hz, 1H), 6.62 (d, J ) 2.3 Hz, 1H), 0.99 (s, 9H), 0.96 (s, 9H), 0.24 (s, 6H), 0.21 (s, 6H); 13C NMR δ 156.9, 153.3, 151.7, 139.9, 131.9, 124.5, 123.9, 121.7, 118.8, 113.7, 113.1, 112.7, 108.7, 90.7, 25.1, 25.1, 17.9, -5.2, -5.2; MS (FD+) m/e 590 (M+). Anal. (C33H42O4SSi2) C, H. (()-3-[(tert-Butyldimethylsilyl)oxy]-6-phenoxy-6H-[1]benzothieno[3,2-c][1]benzopyran (20b). A solution of 15b (1.59 g, 4.1 mmol) and phenol (5.29 g, 56 mmol) in chlorobenzene (50 mL) was heated at reflux for 2.5 h. The mixture was concentrated in vacuo at approximately 70 °C, dissolved in additional chlorobenzene (50 mL), and reconcentrated to yield 1.9 g (100%) of the title compound as a pink solid which was used without further purification: 1H NMR δ 8.05 (d, J ) 8.7 Hz, 1H), 7.85 (d, J ) 8.7 Hz, 1H), 7.3-7.6 (m, 8H), 7.14 (t, J ) 8.2 Hz, 1H), 6.75 (dd, J ) 8.7, 2.1 Hz, 1H), 6.69 (d, J ) 2.1 Hz, 1H), 1.00 (s, 9H), 0.26 (s, 6H). (()-9-(tert-Butyldimethylsilyl)oxy-6-phenoxy-6H-[1]benzothieno[3,2-c][1]benzopyran (20c). The title compound was prepared in 93% yield from 15c by a method similar to that described for 20b: 1H NMR δ 7.76 (d, J ) 8.7 Hz, 1H), 7.60 (d, J ) 2.1 Hz, 1H), 7.56 (d, J ) 8.7 Hz, 1H), 7.1-7.5 (m, 10H), 1.04 (s, 9H), 0.30 (s, 6H). (()-3,9-Dihydroxy-6-[4-[2-(1-piperidinyl)ethoxy]phenyl]6H-[1]benzothieno[3,2-c][1]benzopyran (3a). To a solution of 20a (4.0 g, 6.7 mmol) in toluene (100 mL) at 0 °C was added a 0.5 M THF solution of [4-[2-(1-piperidinyl)ethoxy]phenyl]magnesium bromide (prepared from the corresponding bromobenzene and magnesium turnings, catalyzed by iodine in THF, 27 mL, 13.5 mmol). The mixture was allowed to warm to room temperature and stirred for 1.5 h. After being quenched with water (300 mL), the mixture was extracted with EtOAc (2 × 300 mL), and the organic layer was dried (Na2SO4) and concentrated. The residue was purified via chromatography (silica gel, 3:1 hexane:EtOAc, 0.1% NH4OH) to give 3.85 g (82%) of the title compound as a colorless, gummy solid: 1H NMR δ 7.45 (s, 1H), 7.2-7.3 (m, 4H), 6.8-6.9 (m, 3H), 6.68 (s, 1H), 6.51 (d, J ) 8.1 Hz, 1 H), 6.36 (s, 1H), 4.01 (t, J ) 6.0 Hz, 2H), 2.62 (t, J ) 6.0 Hz, 2H), 2.41 (m, 4H), 1.4-1.6 (m, 4H), 1.3-1.4 (m, 2H), 0.99 (s, 9H), 0.96 (s, 9H), 0.22 (s, 6H), 0.20 (s, 6H); MS (FD) m/e 702 (M+). To a solution of the product of the above reaction (3.85 g, 5.5 mmol) in THF (150 mL) was added a 1.0 M THF solution of TBAF (27.4 mL, 27.4 mmol). The solution was stirred at ambient temperature for 2 h, diluted with EtOAc (300 mL), and washed with saturated NH4Cl (300 mL). The aqueous layer was washed with EtOAc (150 mL), and the combined organic layers were washed with saturated NaHCO3 (300 mL), dried (Na2SO4), and concentrated. The remnant was purified by chromatography (silica gel, 1:1 hexane:EtOAc, 10% MeOH, 0.1% NH4OH) to give 2.35 g (90%) of the titled product as a red foam. Crystallization from MeOH gave an off-white powder, mp 242-245 °C (dec): 1H NMR δ 8.58 (br s, 2H), 7.35 (d, J ) 2.1 Hz, 1H), 7.26 (d, J ) 8.6 Hz, 2H), 7.17 (d, J ) 8.2 Hz, 1H), 7.16 (d, J ) 8.6 Hz, 1H), 6.84 (d, J ) 8.5 Hz, 2H), 6.80 (m, J ) 2.2 Hz, 1H), 6.63 (s, 1H), 6.46 (dd, J ) 8.2, 2.2 Hz, 1H), 6.35 (d, J ) 2.3 Hz, 1H), 4.02 (t, J ) 6.0 Hz, 2H), 2.63 (t, J ) 6.0 Hz, 2H), 2.42 (m, 4H), 1.4-1.6 (m, 4H), 1.31.4 (m, 2H); 13C NMR (DMF-d7) δ 160.2, 159.9, 156.4, 153.2, 140.8, 132.8, 130.9, 129.6, 125.3, 124.6, 122.5, 115.2, 115.1,

1280

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8

Grese et al.

112.2, 109.6, 108.8, 108.7, 104.5, 77.6, 66.6, 58.3, 55.3, 26.5, 24.8; HRMS (FAB+) m/e calcd for C28H28NO4S 474.1739 (MH+), found 474.1726. Anal. (C28H27NO4S‚0.8H2O) C, H, N. (()-3-Hydroxy-6-[4-[2-(1-piperidinyl)ethoxy]phenyl]6H-[1]benzothieno[3,2-c][1]benzopyran (3b). The title compound was prepared in two steps and 82% overall yield from 20b by a method similar to that described for 3a. The product was slurried in MeOH, trifluoroacetic acid was added dropwise until all the material went into solution, the insoluble material was filtered away, and the mother liquor was concentrated in vacuo to yield 0.28 g (80%) of the TFA salt as a fluffy orange solid: 1H NMR δ 7.95 (m, 1H), 7.38 (m, 1H), 7.2-7.3 (m, 5H), 6.90 (d, J ) 8.6 Hz, 2H), 6.76 (s, 1H), 6.51 (dd, J ) 2.2, 8.2 Hz, 1H), 6.38 (d, J ) 2.3 Hz, 1H), 4.44 (t, J ) 4.9 Hz, 2H), 3.5-3.7 (m, 4H), 3.0-3.2 (m, 2H), 1.7-2.0 (m, 5H), 1.4-1.7 (m, 1H); IR (CHCl3) 3271, 3022, 3009, 1670, 1610 cm-1; HRMS (FAB) m/e calcd for C28H28NO3S (MH+) 458.1790, found 458.1781. Anal. (C28H2NO3S‚CF3COOH‚1.2H2O) C, H, N. (()-9-Hydroxy-6-[4-[2-(1-piperidinyl)ethoxy]phenyl]6H-[1]benzothieno[3,2-c][1]benzopyran (3c). The title compound was prepared in two steps and 56% overall yield from 20c by a method similar to that described for 3a: 1H NMR δ 6.7-7.4 (m, 12H), 4.02 (t, J ) 6.0 Hz, 2H), 2.63 (t, J ) 6.0 Hz, 2H), 2.41 (m, 4H), 1.49 (m, 4H), 1.37 (m, 2H); IR (KBr) 2934, 1609 cm-1; HRMS (FAB) m/e calcd for C28H28NO3S (MH+) 458.1790, found 458.1798. Anal. (C28H27NO3S‚0.5H2O) C, H, N. (()-6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-6H-[1]benzothieno[3,2-c][1]benzopyran (3d). To a solution of the 3a (500 mg, 1.06 mmol) in THF (25 mL) containing DMF (2 mL) at room temperature was added Et3N (0.64 g, 0.69 mL, 6.36 mmol) followed by N-phenyltrifluoromethanesulfonimide (0.83 g, 2.33 mmol). The reaction mixture was stirred for 12 h and warmed to 60 °C, and additional N-phenyltrifluoromethanesulfonimide (0.30 g, 0.85 mmol) was added. After 30 min, the reaction mixture was cooled to ambient temperature and concentrated, and the residue was purified by radial chromatography (silica gel, 1:1 hexane:EtOAc, 1% MeOH under an NH3 atmosphere) to afford 780 mg (100%) of the bistriflate as a white foam: 1H NMR (MeOH-d4) δ 7.63 (d, J ) 8.8 Hz, 1H), 7.56 (d, J ) 2.5 Hz, 1H), 7.13 (dd, J ) 8.8, 2.7 Hz, 1H), 7.03 (d, J ) 8.8 Hz, 2H), 6.80 (d, J ) 8.9 Hz, 2H), 6.49 (d, J ) 2.5 Hz, 1H), 6.39 (d, J ) 8.7 Hz, 1H), 6.05 (dd, J ) 8.7, 2.6 Hz, 1H), 4.00 (t, J ) 5.5 Hz, 2H), 2.67 (m, 2H), 2.45 (m, 4H), 1.41.6 (m, 4H), 1.3-1.4 (m, 2H); MS (FD) m/e 737 (M+). A solution of the product of the above reaction (780 mg, 1.06 mmol), Pd(OAc)2 (42 mg, 0.19 mmol), 1,2-bis(diphenylphosphino)propane (149 mg, 0.36 mmol), formic acid (0.6 mL), and Et3N (3.0 mL) in anhydrous DMF (40 mL) was stirred at ambient temperature for 4 d. After concentration, the residue was subjected chromatography (silica gel, 1:1 hexane:EtOAc, 2-10% MeOH, 0.1% NH4OH). Product-containing fractions were concentrated and partitioned between CH2Cl2 (100 mL) and saturated NaHCO3 (100 mL), and the aqueous layer was extracted with CH2Cl2 (50 mL). The combined organic extracts were dried (Na2SO4) and concentrated, and the residue was purified by chromatography (silica gel, 1:1 hexane:EtOAc, 2-10% MeOH, 0.1% NH4OH) to give 267 mg (57%) of the title compound as an oil which gave a white, crystalline solid, mp 107 °C, upon trituration with ether/hexane: 1H NMR (CDCl3) δ 7.86 (d, J ) 7.4 Hz, 1H), 7.40 (m, 1H), 7.1-7.3 (m, 6H), 6.97 (t, J ) 7.3 Hz, 1H), 6.89 (d, J ) 8.1 Hz, 1H), 6.84 (d, J ) 8.6 Hz, 2H), 6.66 (s, 1H), 4.07 (t, J ) 6.1 Hz, 2H), 2.75 (t, J ) 6.1 Hz, 2H), 2.49 (m, 4H), 1.5-1.7 (m, 4H), 1.4-1.5 (m, 2H); 13C NMR (CDCl3) δ 159.1, 151.6, 139.3, 137.1, 133.0, 131.5, 129.8, 129.1, 126.8, 124.6, 124.4, 123.7, 122.6, 121.5, 121.5, 119.3, 116.9, 114.6, 77.6, 65.7, 57.7, 54.9, 25.8, 24.0; MS (FD+) m/e 441 (M+). Anal. (C28H27NO2S) C, H, N. (()-3,9-Dimethoxy-6-[4-[2-(1-piperidinyl)ethoxy]phenyl]6H-[1]benzothieno[3,2-c][1]benzopyran (3e). A solution of 3a (300 mg, 0.633 mmol) in MeOH (75 mL) was treated with a solution of diazomethane in ether/EtOH (approximately 16 mmol) at ambient temperature. The mixture was stirred until

gas evolution ceased, N2 was bubbled through for 10 min to remove excess diazomethane, and the mixture was concentrated. Radial chromatography of the residue (1:1 hexane: EtOAc, 2% MeOH, under an NH3 atmosphere) provided 118 mg (37%) of the title compound as white crystals, mp 134-36 °C: 1H NMR δ 7.51 (d, J ) 2.2 Hz, 1H), 7.2-7.4 (m, J ) 8.6 Hz, 4H), 6.86 (m, 3H), 6.70 (s, 1H), 6.55 (dd, J ) 8.4, 2.3 Hz, 1H), 6.41 (d, J ) 2.4 Hz, 1H), 4.01 (t, J ) 6.0 Hz, 2H), 3.83 (s, 3H), 3.74 (s, 3H), 2.62 (t, J ) 6.0 Hz, 2H), 2.40 (m, 4H), 1.41.6 (m, 4H), 1.3-1.4 (m, 2H); 13C NMR (CDCl3) δ 160.8, 159.2, 157.1, 152.6, 140.3, 131.6, 131.3, 130.5, 129.1, 124.3, 124.0, 121.7, 114.6, 114.0, 112.8, 107.6, 105.7, 102.3, 77.9, 65.8, 57.8, 55.5, 55.2, 54.9, 25.9, 24.1; MS (FD+) m/e 501 (M+). Anal. (C30H31NO4S) C, H, N. (()-3,9-Bis[(tert-butyldimethylsilyl)oxy]-6-[4-hydroxyphenyl]-6H-[1]benzothieno[3,2-c][1]benzopyran (23). To a solution of 20a (3.0 g, 5.08 mmol) in toluene (150 mL) at 0 °C was added a 0.4 M THF solution of [4-(trimethylsilyloxy)phenyl]magnesium bromide (prepared from the corresponding bromobenzene and magnesium turnings, catalyzed by iodine, in THF, 25.4 mL, 10.16 mmol).27 The mixture was allowed to warm to room temperature and stirred for 1.5 h. After dilution with ether (250 mL), the mixture was quenched with saturated NH4Cl (250 mL), and the organic layer was dried (Na2SO4) and concentrated. The residue was slurried in MeOH (100 mL), and ether was added until the mixture was homogeneous. The solution was cooled to 0 °C and treated with anhydrous K2CO3 (3 g) for 15 min. After dilution with ether (250 mL), the mixture was filtered through Celite and washed with saturated NH4Cl, and the aqueous layer was extracted with additional ether (100 mL). The combined organic layers were dried (Na2SO4) and concentrated, and the residue was purified by chromatography (silica gel, 10:1 hexane:EtOAc). Recrystallization from hexane provided 2.6 g (87%) of the title compound as a light pink solid, mp 174-175 °C: 1H NMR δ 8.49 (s, 1H), 7.44 (d, J ) 2.2 Hz, 1H), 7.3-7.4 (m, 4H), 6.83 (dd, J ) 8.7, 2.2 Hz, 1H), 6.76 (d, J ) 8.5 Hz, 2H), 6.65 (s, 1H), 6.50 (dd, J ) 8.2, 2.3 Hz, 1H), 6.36 (d, J ) 2.3 Hz, 1H), 0.98 (s, 9H), 0.95 (s, 9H), 0.21 (s, 6H), 0.20 (s, 6H); 13C NMR δ 158.7, 157.8, 153.9, 153.5, 140.9, 132.7, 131.6, 131.1, 129.9, 126.1, 124.7, 122.7, 119.3, 116.1, 114.4, 114.0, 113.9, 109.3, 78.3, 26.0, 25.9, 18.7, -4.3; IR (CHCl3) 3590, 3310 cm-1; MS (FD+) m/e 590 (M+). Anal. (C33H42O4SSi2) C, H. (()-3,9-Dihydroxy-6-[4-(2-dimethylaminoethoxy)phenyl]-6H-[1]benzothieno[3,2-c][1]benzopyran (3f). A toluene (30 mL) solution of 23 (450 mg, 0.76 mmol), triphenylphosphine (799 mg, 3.1 mmol), and N,N-dimethylethanolamine (340 mg, 383 mL, 3.81 mmol) was treated with DEAD (531 mg, 480 µL, 3.1 mmol) at ambient temperature, and the mixture was stirred for 2 h. The mixture was then diluted with ether (100 mL) and washed with saturated NH4Cl (100 mL), and the aqueous layer was washed with additional ether (50 mL). The combined organic layers were dried (Na2SO4) and concentrated. Chromatography (silica gel, 1:1 hexane: EtOAc, 3% MeOH, 0.1% NH4OH) provided 357 mg (71%) of a white foam: 1H NMR δ 7.45 (d, J ) 2.1 Hz, 1H), 7.27 (d, J ) 8.6 Hz, 2H), 7.24 (m, 2H), 6.83 (m, 3H), 6.67 (s, 1H), 6.50 (dd, J ) 8.2, 2.2 Hz, 1H), 6.36 (d, 2.2 Hz, 1H), 4.00 (t, J ) 5.9 Hz, 2H), 2.60 (t, J ) 5.9 Hz, 2H), 2.20 (s, 6H), 0.98 (s, 9H), 0.95 (s, 9H), 0.21 (s, 6H), 0.20 (s, 6H); MS (FD+) m/e 661.5 (M+). A solution of the above product (328 mg, 0.50 mmol) in THF (10 mL) was treated with 1.0 M TBAF in THF (2.5 mmol) for 2 h, diluted with saturated NH4Cl (100 mL), and extracted with EtOAc (100 mL). The aqueous layer was washed with EtOAc (50 mL), and the combined organic layers were washed with saturated NaHCO3 (2 × 100 mL), dried (Na2SO4), and concentrated. Radial chromatography (silica gel, 1:1 hexane: EtOAc, 30% MeOH, under an NH3 atmosphere) provided 212 mg (99%) of the title compound, which crystallized from CCl4/ acetone as a pink solid, mp 130-140 °C (dec): 1H NMR δ 7.34 (d, J ) 2.1 Hz, 1H), 7.25 (d, J ) 8.6 Hz, 2H), 7.15 (m, 2H), 6.9-7.0 (m, 3H), 6.62 (s, 1H), 6.45 (dd, J ) 8.3, 2.3 Hz, 1H), 6.34 (d, J ) 2.2 Hz, 1H), 4.01 (t, J ) 5.8 Hz, 2H), 2.64 (t, J ) 5.8 Hz, 2H), 2.30 (s, 6H); 13C NMR δ 160.0, 159.8, 156.0, 153.5,

Synthesis of Raloxifene Analogues

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8 1281

141.1, 133.0, 131.4, 130.2, 129.8, 125.3, 124.8, 122.6, 115.3, 115.2, 112.7, 109.7, 109.0, 104.8, 78.0, 66.6, 58.6, 45.8; IR (KBr) 3300 cm-1; HRMS (FAB+) m/e calcd for C25H24NO4S 434.1426 (MH+), found 434.1440. Anal. (C25H23NO4S‚H2O) C, H, N. (()-3,9-Dihydroxy-6-[4-[2-(diethylamino)ethoxy]phenyl]6H-[1]benzothieno[3,2-c][1]benzopyran (3g). The title compound was prepared in two steps and 66% overall yield from 23 and N,N-diethylethanolamine by a method similar to that described for 3f as a red solid, mp 118-123 °C (dec) (acetone/ether): 1H NMR δ 7.35 (d, J ) 2.1 Hz, 1H), 7.26 (d, J ) 8.6 Hz, 2H), 7.16 (d, J ) 8.1 Hz, 1H), 7.13 (d, J ) 8.5 Hz, 1H), 6.81 (m, 3H), 6.62 (s, 1H), 6.45 (dd, J ) 8.3, 2.3 Hz, 1H), 6.35 (d, J ) 2.2 Hz, 1H), 3.98 (t, J ) 6.2 Hz, 2H), 2.78 (t, J ) 6.2 Hz, 2H), 2.56 (q, J ) 7.1 Hz, 4H), 0.97 (t, J ) 7.1 Hz, 6H); 13 C NMR δ 160.2, 159.7, 155.9, 153.5, 141.1, 132.9, 131.5, 130.2, 129.9, 125.4, 124.8, 122.7, 115.2, 112.8, 109.6, 108.9, 104.8, 78.1, 67.5, 52.6, 48.3, 12.4; IR (KBr) 3311 cm-1; MS (FD+) m/e 462 (MH+). Anal. (C27H27NO4S) C, H, N. (()-3,9-Dihydroxy-6-[4-[2-(1-pyrrolidinyl)ethoxy]phenyl]-6H-[1]benzothieno[3,2-c][1]benzopyran (3h). The title compound was prepared in two steps and 70% overall yield from 23 and 1-(2-hydroxyethyl)pyrrolidine by a method similar to that described for 3f as a white solid, mp 237-240 °C (dec) (hexane/EtOAc): 1H NMR (DMF-d7) δ 10.05 (br, 2H), 7.45 (d, J ) 2.0 Hz, 1H), 7.30 (d, J ) 8.4 Hz, 2H), 7.23 (d, J ) 8.6 Hz, 1H), 7.19 (d, J ) 8.2 Hz, 1H), 6.91 (d, J ) 8.7 Hz, 2H), 6.86 (dd, J ) 8.9, 2.2 Hz, 1H), 6.74 (s, 1H), 6.51 (dd, J ) 8.2, 2.1 Hz, 1H), 6.40 (d, J ) 2.1 Hz, 1H), 4.14 (t, J ) 5.6 Hz, 2H), 2.96 (t, J ) 5.5 Hz, 2H), 2.6-2.8 (m, 4H), 1.6-1.8 (m, 4H); 13C NMR (DMF-d7) δ 160.2, 159.6, 156.4, 153.2, 140.8, 133.0, 130.8, 129.6, 125.2, 124.6, 122.4, 115.3, 115.1, 112.2, 109.7, 108.8, 104.5, 77.6, 66.8, 54.8, 54.7, 23.8; IR (KBr) 3286 cm-1; HRMS (FAB+) m/e calcd for C27H26NO4S 460.1583 (MH+), found 460.1572. Anal. (C27H25NO4S‚0.5H2O) C, H, N. (()-3,9-Dihydroxy-6-[4-[2-(1-morpholinyl)ethoxy]phenyl]-6H-[1]benzothieno[3,2-c][1]benzopyran (3i). The title compound was prepared in two steps and 77% overall yield from 23 and 1-(2-hydroxyethyl)morpholine by a method similar to that described for 3f as a red solid, mp 147-153 °C (dec) (acetone/ether): 1H NMR δ 7.35 (d, J ) 2.1 Hz, 1H), 7.25 (d, J ) 8.6 Hz, 2H), 7.16 (d, J ) 8.3 Hz, 1H), 7.14 (d, J ) 8.6 Hz, 1H), 6.81 (m, 3H), 6.62 (s, 1H), 6.46 (dd, J ) 8.2, 2.2 Hz, 1H), 6.35 (d, J ) 2.2 Hz, 1H), 4.04 (t, J ) 5.7 Hz, 2H), 3.56 (t, J ) 4.7 Hz, 4H), 2.68 (t, J ) 5.7 Hz, 2H), 2.47 (t, J ) 4.3 Hz, 4H); 13C NMR δ 160.0, 159.7, 155.9, 153.5, 141.1, 132.9, 131.4, 129.8, 129.7, 125.4, 124.8, 122.6, 115.2, 112.8, 109.6, 108.9, 104.7, 78.0, 67.2, 66.5, 58.1, 54.7; IR (KBr) 3471 cm-1; MS (FD+) m/e 475 (MH+). Anal. (C27H25NO5S‚0.25H2O) C, H, N. (()-3,9-Dihydroxy-6-[4-[2-(2-oxo-1-pyrrolidinyl)ethoxy]phenyl]-6H-[1]benzothieno[3,2-c][1]benzopyran (3j). The title compound was prepared in two steps and 50% overall yield from 23 and 1-(2-hydroxyethyl)-2-pyrrolidinone by a method similar to that described for 3f as a red solid, mp 150160 °C (dec) (acetone solvate): 1H NMR (MeOH-d4) δ 7.21 (m, 3H), 7.11 (d, J ) 8.3 Hz, 1H), 7.02 (d, J ) 8.7 Hz, 1H), 6.80 (d, J ) 8.6 Hz, 2H), 6.71 (dd, J ) 8.6, 2.2 Hz, 1H), 6.51 (s, 1H), 6.37 (dd, J ) 8.2, 2.3 Hz, 1H), 6.25 (d, J ) 2.3 Hz, 1H), 4.04 (t, J ) 5.2 Hz, 2H), 3.57 (t, J ) 5.2 Hz, 2H), 3.48 (t, J ) 7.1 Hz, 2H), 2.28 (t, J ) 8.1 Hz, 2H), 2.14 (s, 6H, CH3CO) 1.91 (quintet, J ) 7.5 Hz, 2H); 13C NMR (MeOH-d4) δ 178.0, 160.2, 160.0, 156.2, 153.9, 141.7, 133.7, 131.9, 130.9, 130.3, 125.6, 125.0, 122.7, 115.6, 115.4, 113.4, 109.8, 109.0, 104.9, 78.6, 66.6, 49.7, 43.4, 31.8, 30.7, 18.9; IR (CHCl3) 1680 cm-1; MS (FD+) m/e 474 (MH+). Anal. (C27H23NO5S‚(CH3)2CO) C, H, N. 3,9-Bis[(tert-butyldimethylsilyl)oxy]-6H-benzofuro[3,2c][1]benzopyran-6-one (5). Coumestrol (10 g, 37.3 mmol) was slurried in CH2Cl2 (600 mL), cooled to 0 °C, and treated with Et3N (24.9 g, 34.3 mL, 246 mmol) and tert-butyldimethylsilyl chloride (24.7 g, 164 mmol). The mixture was warmed to ambient temperature and stirred overnight, during which it slowly became homogeneous. After dilution with ether (800 mL), the mixture was washed with brine (800 mL) and the aqueous layer was extracted with ether (500 mL). The combined organic layers were dried (Na2SO4) and concen-

trated, and the residue was recrystallized from hexane to provide 14.2 g (77%) of the title compound as a white powder, mp 118-119 °C: 1H NMR (CDCl3) δ 7.90 (d, J ) 8.5 Hz, 1H), 7.82 (d, J ) 8.5 Hz, 1H), 7.10 (d, J ) 2.0 Hz, 1H), 6.97 (m, 2H), 6.90 (dd, J ) 8.5, 2.0 Hz, 1H), 1.03 (s, 9H), 1.02 (s, 9H), 0.28 (s, 6H), 0.25 (s, 6H); IR (CHCl3) 1733 cm-1; MS (FD+) m/e 496 (M+). Anal. (C27H36O5Si2) C, H. (()-3,9-Bis[(tert-butyldimethylsilyl)oxy]-6H-benzofuro[3,2-c][1]benzopyran-6-ol (14). The title compound was prepared from 5 in 37% yield as a mixture with its aldehyde tautomer (17) by a method similar to that described for 15a: 1 H NMR δ 10.15 (s, 0.5H), 9.2 (br s, 0.5H), 7.99 (d, J ) 8.4 Hz, 1H), 7.56 (d, J ) 8.4 Hz, 1H), 7.50 (m, 0.5H), 7.09 (d, J ) 2.0 Hz, 1H), 6.96 (m, 1H), 6.6-6.7 (m, 2H), 6.55 (m, 0.5H), 1.01 (s, 9H), 1.00 (s, 9H), 0.27 (s, 6H), 0.25 (s, 6H); IR (CHCl3) 3550, 1663 cm-1; MS (FD) m/e 499 (MH+). Anal. (C27H38O5Si2) C, H. (()-3,9-Bis[(tert-butyldimethylsilyl)oxy]-6-phenoxy6H-benzofuro[3,2-c][1]benzopyran (19). The title compound was prepared in 87% yield from 14 by a method similar to that described for 20a: 1H NMR δ 7.1-7.7 (m, 8H) 6.6-6.9 (m, 3H), 1.01 (s, 9H), 1.00 (s, 9H), 0.28 (s, 6H), 0.25 (s, 6H). (()-3,9-Dihydroxy-6-[4-[2-(1-piperidinyl)ethoxy]phenyl]6H-benzofuro[3,2-c][1]benzopyran (2). The title compound was prepared in two steps and 65% overall yield from 19 by a method similar to that described for 3a as a bright red foam, which crystallized from CCl4: 1H NMR (DMF-d7) δ 10.00 (br s, 1H), 9.83 (br s, 1H), 7.3-7.5 (m, 3H), 7.10 (s, 1H), 6.98 (d, J ) 8.5 Hz, 2H), 6.7-6.9 (m, 3H), 6.54 (dd, J ) 8.1, 1.8 Hz, 1H), 6.42 (s, 1H), 4.10 (t, J ) 5.8 Hz, 2H), 2.67 (m, 2H), 2.44 (m, 4H), 1.4-1.6 (m, 4H), 1.3-1.4 (m, 2H); 13C NMR (DMFd7) δ 160.2, 159.9, 156.8, 156.4, 155.1, 147.2, 133.0, 129.5, 121.4, 119.5, 118.9, 115.1, 112.9, 109.2, 108.9, 108.3, 104.3, 98.7, 78.5, 66.5, 58.2, 55.3, 26.4, 24.6; IR (KBr) 3220 cm-1; HRMS (FAB+) m/e calcd for C28H28NO5 458.1967 (MH+), found 458.1974. Anal. (C28H27NO4S‚1.75H2O) C, N; H: calcd, 6.30; found, 5.81. (()-2-Methoxy-8-hydroxy-11,12-dihydro-5H-benzo[b]naphtho[2,1-d]pyran-5-one (13). To a solution of 1-carbomethoxy-6-methoxy-2-tetralone (12)27 (18.0 g, 76.8 mmol) and resorcinol (8.9 g, 80.7 mmol) being stirred at ambient temperature in toluene (450 mL) was added POCl3 (12.0 g, 7.3 mL, 18.3 mmol) dropwise, and the mixture was warmed to 80 °C for 12 h. After being cooled to room temperature, the mixture was poured into water (500 mL) and filtered, and the precipitate was rinsed with ether. The filtrate layers were separated, the aqueous layer was extracted with EtOAc (3 × 500 mL), and the combined organic layers were dried (Na2SO4) and concentrated. Recrystallization of the residue from MeOH provided 16.0 g (71%) of the title compound as a yellow solid, mp 244-249 °C (dec): 1H NMR (DMSO-d6) δ 10.55 (br s, 1H), 8.24 (d, J ) 8.6 Hz, 1H), 7.75 (d, J ) 8.7 Hz, 1H), 6.67.0 (m, 4H), 3.76 (s, 3H), 2.8-3.0 (m, 4H); 13C NMR (DMSOd6) δ 170.3, 160.3, 158.3, 158.1, 153.3, 147.7, 137.5, 127.4, 125.8, 122.5, 114.5, 112.6, 112.4, 110.9, 101.4, 54.6, 26.3, 22.7; IR (KBr) 3250, 1676, 1618 cm-1; MS (FD+) m/e 294 (M+). Anal. (C18H14O4) C, H. 2,8-Bis[(tert-butyldimethylsilyl)oxy]-5H-benzo[b]naphtho[2,1-d]pyran-5-one (7). To a mechanically stirred slurry of 13 (4.17 g, 14.2 mmol) in CH2Cl2 (100 mL) was added ethanethiol (3.53 g, 3.95 mL, 56.8 mmol) followed by aluminum chloride (9.0 g, 67.5 mmol), portionwise. The reaction mixture was stirred at ambient temperature for 5 h, cooled to 0 °C, and quenched cautiously with THF (100 mL) followed by saturated NaHCO3 (100 mL). The mixture was diluted with THF (100 mL), the layers were separated, and the aqueous layer was washed with THF (100 mL). The combined organic layers were dried (Na2SO4) and concentrated to yield 3.6 g of crude diphenol as a yellow solid, which was used without further purification. The crude product was slurried in CH2Cl2 (100 mL) and treated with Et3N (8.6 g, 11.9 mL, 85.2 mmol) and tertbutyldimethylsilyl chloride (8.6 g, 56.8 mmol). The mixture was stirred at ambient temperature for 5 h, during which it

1282

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8

Grese et al.

slowly became homogeneous. After dilution with ether (250 mL), the mixture was washed with brine (250 mL) and the aqueous layer was extracted with ether (150 mL). The combined organic layers were dried (Na2SO4) and concentrated, and the residue was recrystallized from hexane to provide 5.3 g (74%) of the title compound as a yellow solid, mp 145-147 °C: 1H NMR (CDCl3) δ 8.37 (d, J ) 8.6 Hz, 1H), 7.58 (d, J ) 9.3 Hz, 1H), 6.83 (s, 1H), 6.81 (m, 2H), 6.72 (d, J ) 2.4 Hz, 1H), 2.8-3.0 (m, 4H), 1.00 (s, 18H), 0.27 (s, 6H), 0.24 (s, 6H); MS (FD) m/e 508 (M+). A sample of the above product (7.0 g, 13.8 mmol) and DDQ (3.3 g, 14.5 mmol) in DCE (100 mL) was heated at reflux overnight, inducing the formation of a precipitate. The mixture was cooled to room temperature, diluted with hexane (300 mL), filtered through Celite, and concentrated. The remnant was partitioned between hexane:ether (1:1, 200 mL) and water (200 mL), and the organic layer was dried (MgSO4), concentrated, and recrystallized from hexane to provide 5.5 g (79%) of the title product as a tan solid, mp 154-156 °C: 1H NMR (CDCl3) δ 9.69 (d, J ) 9.4 Hz), 8.80 (s, 2H), 8.01 (d, J ) 9.5 Hz), 7.34 (dd, J ) 9.3,2.5 Hz, 1H), 7.28 (d, J ) 5.4 Hz, 1H), 6.90 (m, 2H), 1.05 (s, 9H), 1.03 (s, 9H), 0.29 (s, 12H); IR (CHCl3) 1711, 1622, 1604 cm-1; MS (FD) m/e 506 (M+). Anal. (C29H38O4Si2) C, H. (()-2,8-Bis[(tert-butyldimethylsilyl)oxy]-5H-benzo[b]naphtho[2,1-d]pyran-5-ol (16). The title compound was prepared from 7 in 82% yield by a method similar to that described for 15a as a white solid, mp 188-190 °C (hexane/ ether): 1H NMR (CDCl3) δ 7.98 (d, J ) 9.1 Hz, 1H), 7.86 (d, J ) 8.8 Hz, 1H), 7.79 (d, J ) 8.6 Hz, 1H), 7.75 (d, J ) 8.2 Hz, 1H), 7.23 (d, J ) 2.3 Hz, 1H), 7.18 (dd, J ) 9.0, 2.4 Hz, 1H), 7.06 (d, J ) 7.4 Hz, 1H), 6.67 (m 2H), 3.24 (d, J ) 7.7 Hz, 1H), 1.04 (s, 9H), 1.02 (s, 9H), 0.27 (s, 6H), 0.27 (s, 6H); IR (CHCl3) 3574 cm-1; MS (FD) m/e 508 (M+). Anal. (C29H40O4Si2) Calcd: C, 68.44; H, 7.94. Found: C, 68.63; H, 8.11. (()-2,8-Bis[(tert-butyldimethylsilyl)oxy]-5-phenoxy5H-benzo[b]naphtho[2,1-d]pyran (21). The title compound was prepared in 95% yield from 16 by a method similar to that described for 20a: 1H NMR δ 7.9-8.1 (m, 4H), 7.61 (s, 1H), 7.3-7.5 (m, 3H), 7.2-7.3 (m, 3H), 7.11 (t, J ) 7.3 Hz, 1H), 6.74 (dd, J ) 8.5, 2.3 Hz, 1H), 6.62 (d, J ) 2.4 Hz, 1H), 1.03 (s, 9H), 0.99 (s, 9H), 0.29 (s, 6H), 0.24 (s, 6H); MS (FD) m/e 584 (M+). (()-2,8-Dihydroxy-5-[4-[2-(1-piperidinyl)ethoxy]phenyl]5H-benzo[b]naphtho[2,1-d]pyran (4). The title compound was prepared in two steps and 81% overall yield from 21 by a method similar to that described for 3a as a white solid, mp 158-161 °C (ether): 1H NMR δ 7.89 (d, J ) 8.7 Hz, 1H), 7.77 (d, J ) 8.7 Hz, 1H), 7.68 (m, 2H), 7.23 (d, J ) 2.4 Hz, 1H), 7.09 (m, 3H), 6.97 (s, 1H), 6.71 (d, J ) 8.7 Hz, 2H), 6.49 (dd, J ) 8.3, 2.4 Hz, 1H), 6.36 (d, J ) 2.3 Hz, 1H), 3.95 (t, J ) 6.0 Hz, 2H), 2.60 (t, J ) 6.0 Hz, 2H), 2.40 (m, 4H), 1.4-1.5 (m, 4H), 1.3-1.4 (m, 2H); 13C NMR δ 159.5, 159.5, 155.8, 153.9, 135.0, 132.4, 130.1, 128.0, 126.7, 125.6, 125.4, 125.3, 124.7, 121.4, 119.9, 116.1, 114.8, 111.0, 110.1, 105.5, 75.8, 66.1, 58.3, 55.3, 26.3, 24.7; IR (KBr) 2934 cm-1; MS (FD) m/e 468 (MH+). Anal. (C30H29NO4‚H2O) C, H, N. Biological Assays. Methods utilized for the ER binding, MCF-7 proliferation, 4-day and 5-week OVX rat assays have been recently described.35 Procedures for the immature rat assay have also been published elsewhere.45 Stimulation of TGF-β3 promoter activity was assayed in HeLa cells which had been cotransfected with a TGF-β3/luciferase reporter plasmid and an expression plasmid (pCMVER) encoding the human ER by the published procedure.43

of Lilly Research Laboratories (analytical and spectral data and support).

Acknowledgment. The authors thank Dr. Chris Schmid for helpful discussions and suggestions. The authors also thank Kennan Fahey, Dr. David Mitchell, and Robert Murff for the preparation of starting materials and intermediates. The authors also acknowledge the contributions of the Physical Chemistry Department

References (1) Lindsay, R. Sex steroids in the pathogenesis and prevention of osteoporosis. In Osteoporosis: Etiology, Diagnosis, and Management; Riggs, B. L., Melton, L. J., Eds.; Raven: New York, 1988; pp 333-358. (2) Ross, R. K.; Pike, M. C.; Mack, T. M.; Henderson, B. E. Oestrogen replacement therapy and cardiovascular disease. In HRT and Osteoporosis; Drife, J. O., Studd, J. W. W., Eds.; SpringerVerlag: London, 1990; pp 209-222. (3) Cust, M. P.; Gangar, K. F.; Whitehead, M. I. Oestrogens and cancer. In HRT and Osteoporosis; Drife, J. O., Studd, J. W. W., Eds.; Springer-Verlag: London, 1990; pp 177-186. (4) Session, D. R.; Kelly, A. C.; Jewelewicz, R. Current concepts in estrogen replacement therapy in the menopause. Fertility Sterility 1993, 2, 277-284 and references therein. (5) (a) Colditz, G. A.; Hankinson, S. E.; Hunter, D. J.; Willett, W. C.; Manson, J. E.; Stampfer, M. J.; Hennekens, C.; Rosner, B.; Speizer, F. E. The use of estrogens and progestins and the risk of breast cancer in postmenopausal women. N. Engl. J. Med. 1995, 332, 1589-1593. (b) Collaborative Group on Hormonal Factors in Breast Cancer. Breast cancer and hormone replacement therapy: Collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women withou breast cancer. Lancet 1997, 350, 10471059. (6) Beresford, S. A. A.; Weiss, N. S.; Voigt, L. F.; McKnight, B. Risk of endometrial cancer in relation to use of oestrogen combined with cyclic progestagen therapy in postmenopausal women. Lancet 1997, 349, 458-461. (7) For recent reviews, see: (a) Miquel, J.-F.; Gilbert, J. A chemical classification of nonsteroidal antagonists of sex-steroid hormone action. J. Steroid Biochem. Mol. Biol. 1988, 31, 525-544. (b) Magarian, R. A.; Overacre, L. B.; Singh, S.; Meyer, K. L. The medicinal chemistry of nonsteroidal antiestrogens. Curr. Med. Chem. 1994, 1, 61-104. (c) Chander, S. K.; Sahota, S. S.; Evans, T. R. J.; Luqmani, Y. A. The biological evaluation of novel antioestrogens for the treatment of breast cancer. Crit. Rev. Oncol. Hematol. 1993, 15, 243-269. (d) Jordan, V. C. Alternate antiestrogens and approaches to the prevention of breast cancer. J. Cell Biochem. 1995, 51-57. (8) Jordan, V. C. A current view of tamoxifen for the treatment and prevention of breast cancer. Br. J. Pharmacol. 1993, 110, 507517. (9) For recent reviews, see: (a) Grese, T. A.; Dodge, J. A. Estrogen receptor modulators: effects in non-traditional target tissues. Annu. Rep. Med. Chem. 1996, 31, 181-190. (b) Kauffman, R. F.; Bryant, H. U. Selective estrogen receptor modulators. Drug News Perspect. 1995, 8, 531-539. (c) Evans, G. L.; Turner, R. T. Tissue-selective actions of estrogen analogs. Bone 1995, 17, 181S-190S. (10) Jones, C. D.; Jevnikar, M. G.; Pike, A. J.; Peters, M. K.; Black, L. J.; Thompson, A. R.; Falcone, J. F.; Clemens, J. A. Antiestrogens. 2. Structure-activity studies in a series of 3-aroyl-2arylbenzo[b]thiophene derivatives leading to [6-hydroxy-2-(4hydroxyphenyl)benzo[b]thien-3-yl][4-[2-(1-piperidinyl)ethoxy]phenyl]methanone hydrochloride (LY156758), a remarkably effective estrogen antagonist with only minimal intrinsic estrogenicity. J. Med. Chem. 1984, 27, 1057-1066. (11) (a) Draper, M. W., Flowers, D. E.; Huster, W. J.; Neild, J. A.; Harper, K. D.; Arnaud, C. A controlled trial of raloxifene (LY139481) HCl: Impact on bone turnover and serum lipid profile in healthy postmenopausal women. J. Bone Miner. Res. 1996, 11, 835-842. (b) Delmas, P. D.; Bjarnason, N. H.; Mitlak, B. H.; Ravoux, A.-C.; Shah, A. S.; Huster, W. J.; Draper, M.; Christiansen, C. Effects of raloxifene on bone mineral density, serum cholesterol concentrations, and uterine endometrium in postmenopausal women. N. Engl. J. Med. 1997, 337, 1641. (12) (a) Sato, M.; Rippy, M. K.; Bryant, H. U. Raloxifene, tamoxifen, nafoxidine, or estrogen effects on reproductive and nonreproductive tissues in ovariectomized rats. FASEB J. 1996, 10, 905912. (b) Black, L. J.; Sato, M.; Rowley, E. R.; Magee, D. E.; Bekele, A.; Williams, D. C.; Cullinan, G. J.; Bendele, R.; Kauffman, R. F.; Bensch, W. R.; Frolik, C. A.; Termine, J. D.; Bryant, H. U. Raloxifene (LY139481 HCl) prevents bone loss and reduces serum cholesterol without causing uterine hypertrophy in ovariectomized rats. J. Clin. Invest. 1994, 93, 63-69. (13) Another group has recently described proliferative effects in uterine tissue from immature rats treated with raloxifene: Ashby, J.; Odum, J.; Foster, J. R. Activity of raloxifene in immature and ovariectomized rat uterotrophic assays. Regul. Toxicol. Pharm. 1997, 25, 226-231. Our data from both immature and ovariectomized rats, reported herein and elsewhere,12,18,34 do not support their findings or conclusions.

Synthesis of Raloxifene Analogues (14) Gauthier, S.; Caron, B.; Cloutier, J.; Dory, Y. L.; Favre, A/; Larouche, D.; Mailhot, J.; Ouellet, C.; Schwerdtfeger, A.; Leblanc, G.; Martel, C.; Simard, J.; Me´rand, Y.; Be´langer, A.; Labrie, C.; Labrie, F. (S)-(+)-4-[7-(2,2-Dimethyl-1-oxopropoxy)-4-methyl2-[4-[2-(1-piperidinyl)ethoxy]phenyl]-2H-1-benzopyran-3-yl]-phenyl 2,2-dimethylpropanoate (EM-800): A highly potent, specific, and orally active nonsteroidal antiestrogen. J. Med. Chem. 1997, 40, 2117-2122. (15) Sharma, A. P.; Saeed, A.; Durani, S.; Kapil, R. S. Structureactivity relationship of antiestrogens. Phenolic analogues of 2,3diaryl-2H-1-benzopyrans. J. Med. Chem. 1990, 33, 3222 and references therein. (16) Grese, T. A.; Sluka, J. P.; Bryant, H. U.; Cole, H. W.; Kim, J. R.; Magee, D. E.; Rowley, E. R.; Sato, M. Benzopyran selective estrogen receptor modulators (SERMs): pharmacological effects and structural correlation with raloxifene. Bioorg. Med. Chem. Lett. 1996, 6, 903-908. (17) (a) Boyd, D. B.; Coner, R. D. Stochastic approach to force field evaluations: conformational analysis of raloxifene, a potential new therapeutic agent for post-menopausal osteoporosis. J. Mol. Struct. (THEOCHEM) 1996, 368, 7-15. (b) Kym, P. R.; Anstead, G. M.; Pinney, K. G.; Wilson, S. R.; Katzenellenbogen, J. A. Molecular structures, conformation analysis, and preferential modes of binding of 3-aroyl-2-arylbenzo[b]thiophene estrogen receptor ligands: LY117018 and aryl azide photoaffinity labeling analogs. J. Med. Chem. 1993, 36, 3910-3922. (c) Brzozowski, A. M.; Pike, A. C. W.; Dauter, Z.; Hubbard, R. E.; Monn, T.; Engstro¨m, O.; O ¨ hman, L.; Greene, G. L.; Gustafsson, J.-Å.; Carlquist, M. Molecular basis of agonism and antagonism in the oestrogen receptor. Nature 1997, 389, 753-757. (18) Grese, T. A.; Sluka, J. P.; Bryant, H. U.; Cullinan, G, J.; Glasebrook, A. L.; Jones, C. D.; Matsumoto, K.; Palkowitz, A. D.; Sato, M.; Termine, J. D.; Winter, M. A.; Yang, N. N.; Dodge, J. A. Molecular determinants of selectivity in estrogen receptor modulators. Proc. Natl. Acad. Sci. U.S.A. 1997, 94, 1410514110. (19) Representative low-energy conformations were generated using the AM1 parameter set with the MOPAC20 command line options of AM1 GRADIENT POLAR GEO-OK HESS)1 EF GNORM)0.01SCFCRT)1D-12 MMOK PRECISE NOINTER and GRAPH. Least squares superimpositions of the stilbene cores were performed using the Insight-II version 95.0.4 (Molecular Simulations Inc., San Diego, CA). (20) By James P. Stewart, Frank J. Seller, Research Laboratory, U.S. Air Force Academy, CO 90940. QCPE No. 455 and references therein. (21) Grese, T. A.; Pennington, L. D. Novel methodology for the synthesis of estrogenic and antiestrogenic isoflav-3-enes. Tetrahedron Lett. 1995, 36, 8913-8916. (22) A portion of this work has previously appeared in communication form: Grese, T. A.; Cole, H. W.; Magee, D. E.; Phillips, D. L.; Shetler, P. K.; Short, L. L.; Glasebrook, A. L.; Bryant, H. U. Conversion of the phytoestrogen coumestrol into a selective estrogen receptor modulator (SERM) by attachment of an aminecontaining side chain. Bioorg. Med. Chem. Lett. 1996, 6, 26832686. (23) Conley, R. A.; Heindel, N. D. Thianaphthen-2-one chemistry. I. Synthesis of 6H-benzothieno[3,2-c][1]benzopyran-6-ones (11thiacoumestans). J. Org. Chem. 1975, 40, 3169-3173. (24) Ablenas, F. J.; George, B. E.; Maleki, M.; Jain, R.; Hopkinson, A. C.; Lee-Ruff, E. Destabilized carbocations. Nuclear magnetic resonance detection and reactivities of aryl R-thioformamidyl cations. Can. J. Chem. 1987, 65, 1800-1803. (25) Schmid, C. R., unpublished results. (26) Lumma, W. C., Jr.; Dutra, G. A.; Voekker, C. A. Synthesis of two benzothiacyclanones via a novel two-carbon ring expansion of thiolactones with vinyllithium. J. Org. Chem. 1970, 35, 34423444. (27) Colvin, E. W.; Martin, J.; Shroot, B. Selective carboalkoxylation of 6-methoxy-2-tetralone. Chem. Ind. (London) 1966, 2130. (28) Sethna, S.; Phadke, R. The Pechman reaction. Org. React. 1953, 7, 1-58. (29) Bindal, R. D.; Durani, S.; Kapil, R. S.; Anand, N. Utility of 4-trimethylsiloxyphenylmagnesium bromide in Grignard reactions. Synthesis 1982, 405. (30) Glasebrook, A. L.; Phillips, D. L.; Sluka, J. P. Multiple binding sites for the anti-estrogen raloxifene (LY156758). J. Bone Miner. Res. 1993, 8 (Suppl. 1), S268. Scholl, S. M.; Huff, K. K.; Lippman, M. E. Antiestrogenic effects of LY117018 in MCF-7 cells. Endocrinology 1983, 113, 611-617. Miller, M. A.; Katzenellenbogen, B. S. Characterization and quantitation of antiestrogen binding sites in estrogen receptor-positive and -negative human breast cancer cell lines. Cancer Res. 1983, 43, 3094-3100. (31) Thompson, E. W.; Reich, R.; Shima, T. B.; Albini, A.; Graf, J.; Martin, G. R.; Dickson, R. B.; Lippman, M. E. Differential growth and invasiveness of MCF-7 breast cancer cells by antiestrogens. Cancer Res. 1988, 48, 6764-6768.

Journal of Medicinal Chemistry, 1998, Vol. 41, No. 8 1283 (32) Bryant, H. U.; Bendele, A. M.; Magee, D. E.; Cole, H. W.; Spaethe, S. W. Pharmacologic profile of estrogen-induced uterine eosiophilia in the ovariectomized (OVX) rat. Pharmacologist 1992, 34, 194. (33) Wronski, T. J.; Yen, C.-F. The ovariectomized rat as an animal model for postmenopausal bone loss. Cells Mater. 1991, Suppl 1, 69. (34) (a) Fuchs-Young, R.; Magee, D. E.; Cole, H. W.; Short, L.; Glasebrook, A. L.; Rippy, M. K.; Termine, J. D.; Bryant, H. U. Raloxifene is a tissue specific anti-estrogen that blocks tamoxifen or estrogen-stimulated uterotrophic effects. Endocrinology 1995, 135 (Suppl.), 57. (b) Bryant, H. U.; Wilson, P. K.; Adrian, M. D.; Cole, H. W.; Short, L. L.; Dodge, J. A.; Grese, T. A.; Sluka, J. P.; Glasebrook, A. L. Selective estrogen receptor modulators: Pharmacological profile in the rat uterus. J. Soc. Gynecol. Invest. 1996, 3(Suppl), 152A. (35) Grese, T. A.; Cho, S.; Finley, D. R.; Godfrey, A. G.; Jones, C. D.; Lugar, C. W., III.; Martin, M. J.; Matsumoto, K.; Pennington, L. D.; Winter, M. A.; Adrian, M. D.; Cole, H. W.; Magee, D. E.; Phillips, D. L.; Rowley, E. R.; Short, L. L.; Glasebrook, A. L.; Bryant, H. U. Structure-activity relationships of selective estrogen receptor modulators: modifications to the 2-arylbenzothiophene core of raloxifene. J. Med. Chem. 1997, 40, 146167. (36) Anstead, G. M.; Carlson, K. E.; Katzenellenbogen, J. A. The estradiol pharmacophore: ligand structure-estrogen receptor binding affinity relationships and a model for the receptor binding site. Steroids 1997, 62, 268-303. (37) Dodge, J. A. Presented at the 25th National Medicinal Chemistry Symposium, Ann Arbor, MI, June 1996. (38) On the other hand, in the tamoxifen series, substitution of the dimethylamino group by piperidine does not result in a reduction of uterine stimulation: Robertson, D. W.; Katzenellenbogen, J. A.; Hayes, J. R.; Katzenellenbogen, B. S. J. Med. Chem. 1982, 25, 167-171. Therefore, the presence of cyclic amino functionality appears to be necessary but not sufficient to achieve slectivity with respect to uterine effects (ref 18). (39) Rosati, R. L.; Da Silva Jardine, P.; Cameron, K. O.; Thompson, D. D.; Ke, H. Z.; Toler, S. M.; Brown, T. A.; Pan, L. C.; Ebbinghaus, C. F.; Reinhold, A. R.; Elliot, N. C.; Newhouse, B. N.; Tjoa, C. M.; Sweetnam, P. M.; Cole, M. J.; Arriola, M. W.; Gauthier, J. W.; Crawford, D. T.; Nickerson, D. F.; Pirie, C.; Qi, H.; Simmons, H. A.; Tkalcevic, G. T. Discovery and SAR of a potent nonsteroidal estrogen agonist CP-336,156. J. Bone Miner. Res. 1996, 11 (Suppl. 1), S447. (40) Dodge, J. A.; Lugar, C. W.; Cho, S.; Osborne, J. J.; Phillips, D. L.; Glasebrook, A. L.; Frolik, C. A. Synthesis and estrogen receptor binding affinities of the major human metabolites of raloxifene (LY139481). Bioorg. Med. Chem. Lett. 1997, 7, 993996, and references therein. (41) (a) McDonnell, D. P.; Clemm, D. L.; Hermann, T.; Goldman, M. E.; Pike, J. W. Analysis of estrogen receptor function in vitro reveals three distinct classes of antiestrogens. Mol. Endocrinol. 1995, 9, 659-669. (b) Klinge, C. M.; Traish, A. M.; Driscoll, M. D.; Hilf, R.; Bambara, R. A. Site-directed estrogen receptor antibodies stabilize 4-hydroxytamoxifen ligand, but not estradiol, and indicate ligand-specific differences in the recognition of estrogen response element DNA in vitro. Steroids 1996, 61, 278289. (42) For a detailed description of the current understanding of tissuespecific steroid hormone activities, see: Katzenellenbogen, J. A.; O’Malley, B. W.; Katzenellenbogen, B. S. Tripartite steroid hormone receptor pharmacology: Interaction with multiple effector sites as a basis for the cell- and promoter-specific action of these hormones. Mol. Endocrinol. 1996, 10, 119-131. (43) (a) Yang, N. N.; Venugopalan, M.; Hardikar, S.; Glasebrook, A. G. Identification of an estrogen response element activated by metabolites of 17β-estradiol and raloxifene. Science 1996, 273, 1222-1225. (b) Yang, N. N.; Bryant, H. U.; Hardikar, S.; Sato, M.; Galvin, R. J. S.; Glasebrook, A. L.; Termine, J. D. Estrogen and raloxifene stimulate transforming growth factor-β3 gene expression in rat bone: a potential mechanism for estrogen- or raloxifene-mediated bone maintenance. Endocrinology 1996, 137, 2075-2084. (44) Salman, M.; Ray, S.; Anand, N.; Agarwal, A. K.; Singh, M. M.; Setty, B. S.; Kamboj, V. P. Studies in antifertility agents. 50: Stereoselective binding of d- and l-centchromans to estrogen receptors and their antifertility activity. J. Med. Chem. 1986, 29, 1801-1803. (45) Palkowitz, A. D.; Glasebrook, A. L.; Thrasher, K. J.; Hauser, K. L.; Short, L. L.; Phillips, D. L.; Muehl, B. S.; Sato, M.; Shetler, P. K.; Cullinan, G. J.; Pell, T. R.; Bryant, H. U. Discovery and synthesis of [6-hydroxy-3-[4-[2-(1-piperidinyl)ethoxy]phenoxy]2-(4-hydroxyphenyl)]benzo[b]thiophene: A novel, highly potent, selective estrogen receptor modulator. J. Med. Chem. 1997, 10, 11407-1416.

JM970688Z